Next Article in Journal
Sorbitol as a Chain Extender of Polyurethane Prepolymers to Prepare Self-Healable and Robust Polyhydroxyurethane Elastomers
Next Article in Special Issue
Phytochemical Constituents and Biological Activities of Melicope lunu-ankenda
Previous Article in Journal
Optimization of Ultrasound-Assisted Extraction of Flavonoids from Olive (Olea europaea) Leaves, and Evaluation of Their Antioxidant and Anticancer Activities
Previous Article in Special Issue
Trichosanthis Pericarpium Aqueous Extract Protects H9c2 Cardiomyocytes from Hypoxia/Reoxygenation Injury by Regulating PI3K/Akt/NO Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

UPLC-MS/MS Method for the Determination of 14 Compounds in Rat Plasma and Its Application in a Pharmacokinetic Study of Orally Administered Xiaoyao Powder

1
Key Laboratory of Photochemistry Biomaterials and Energy Storage Materials of Heilongjiang Province, College of Chemistry & Chemical Engineering, Harbin Normal University, No.1 South Road, Limin Development Zone, Harbin 150025, China
2
College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, Heilongjiang 154007, China
3
Crop Academy of Heilongjiang University, Harbin 150080, China
*
Authors to whom correspondence should be addressed.
Molecules 2018, 23(10), 2514; https://doi.org/10.3390/molecules23102514
Submission received: 9 September 2018 / Revised: 25 September 2018 / Accepted: 26 September 2018 / Published: 30 September 2018
(This article belongs to the Special Issue Natural Product Pharmacology and Medicinal Chemistry)

Abstract

:
Xiaoyao Powder (XYP), a common Chinese medicine, comprises eight traditional Chinese herbs and has been widely used clinically to treat liver damage and mental disorders. An ultra-performance liquid chromatography–tandem mass spectrometry method was developed to investigate the pharmacokinetics of 14 compounds (albiflorin, paeoniflorin, ferulic acid, senkyunolide I, quercetin, isoliquiritigenin, atractylenolide III, ligustilide, atractylenolide II, liquiritin, liquiritigenin, saikosaponin c, glycyrrhizic acid, and saikosaponin a) in XYP. Naringenin was used as the internal standard. The compounds were separated using an ACQUITY UPLCTM BEH C18 column (1.7 μm, 50 × 2.1 mm) with a mobile phase consisting of acetonitrile and 0.1% formic acid in water at a flow rate of 0.3 mL/min. Detection was performed on a triple-quadrupole tandem mass spectrometer using multiple reaction monitoring and an electrospray ionization source in both positive and negative ionization modes. All calibration curves exhibited good linearity (r2 > 0.9974) over the measured ranges. The intra- and inter-day precisions were within 12%, and the accuracy ranged from 89.93% to 106.64%. Extraction recovery and matrix effect results were satisfactory. The method was successfully applied in a pharmacokinetic study of the 14 compounds in rat plasma after the oral administration of XYP.

Graphical Abstract

1. Introduction

Xiaoyao Powder (XYP), a traditional Chinese medicine formula, was first described in the Taiping Huimin Heji Jufang during the Song Dynasty of China (960–1127 AD). This formula has been widely used to treat liver damage and as an antidepressant for more than 2000 years [1,2,3,4]. According to the Chinese Pharmacopoeia, XYP consists of Radix Bupleuri (Bupleurum chinense DC), Radix Angelicae Sinensis (Angelica sinensis (Oliv.) Diels), Radix Paeoniae Alba (Paeonia lactiflora Pall.), Rhizoma Atractylodis Macrocephalae (Atractylodes macrocephala Koidz.), Poria (Poria cocos (Schw.) Wolf), Radix Glycyrrhizae (Glycyrrhiza uralensis Fisch.), Herba Menthae (Mentha haplocalyx Briq.) and Rhizoma Zingiberis Recens (Zingiber officinale Rosc.) in a ratio of 5:5:5:5:5:4:1:1 [5].
In our previous study, the components of XYP were identified by ultra-performance liquid chromatography coupled with electrospray ionization tandem mass spectrometry (UPLC-ESI-MS) and gas chromatography-mass spectrometry (GC-MS), yielding a comprehensive list of compounds in XYP [6,7]. XYP and these principle constituents showed sufficient hepatoprotective effects on hepatic fibrosis induced in rats by CCl4 [8]. Pharmacokinetics is helpful to evaluate the rationality and compatibility of Chinese herbal medicine or prescription, in terms of efficacy and toxicity [9]. Up to now, there are no reports on which of these XYP compounds are absorbed, nor which may have a key role in the pharmacokinetics of XYP.
Firstly, a serum pharmacochemistry study was performed, where we tried to determine the components’ plasma absorption after oral administration of XYP to rats. In the pilot study, 14 compounds were identified by UPLC-MS/MS as chemical entities that were absorbed from the gastrointestinal tract after oral administration. Some of these compounds have been reported to be related with hepatoprotective or antidepressant effects. Radix Bupleuri is a principal herb in this formula for liver disorder treatment, and quercetin, saikosaponin a and saikosaponin c from this herb were proved to be the effective ingredients [10,11,12]. Bioactivity studies showed that ligustilide is the major compound in Radix Angelicae Sinensis connected with central nervous system protection [13,14]. As another main compound of Radix Angelicae Sinensis, ferulic acid showed protective activities against CCl4-induced liver injury in mice [15]. Liquiritin, liquiritigenin, isoliquiritigenin, and glycyrrhizic acid are the major hepatoprotective components of Radix Glycyrrhizae [16,17,18,19,20]. Paeoniflorin and albiflorin are the most abundant compounds in Radix Paeoniae Alba [21,22,23]. Senkyunolide I can protect nervus centralis against focal cerebral ischemia-reperfusion injury [24]. Rhizoma Atractylodis Macrocephalae has been used in traditional Chinese medicine to treat melancholia. Its main active components are atractylenolide II and atractylenolide III [25,26]. Based on the pilot study described above and our literature review, we determined to study the pharmacokinetic characteristics of these 14 compounds (albiflorin, paeoniflorin, ferulic acid, senkyunolide I, quercetin, isoliquiritigenin, atractylenolide III, ligustilide, atractylenolide II, liquiritin, liquiritigenin, saikosaponin c, glycyrrhizic acid, and saikosaponin a) that are most likely to represent the pharmacokinetic profile of XYP.
Pharmacokinetics studies of some of these 14 compounds in other Chinese formula or single herb have been reported. However, the possibility of interaction between components in an herbal formula and the pharmacokinetic variation of one single compound in different formulas that a multiple-components pharmacokinetic study would be more likely to illustrate the pharmacokinetic profile of XYP. An ultra-performance liquid chromatography–tandem mass spectrometry (UPLC-MS/MS) method is the best and high efficient approach for simultaneous determination of multiple compounds in a complex matrix [27]. Thus, the pharmacokinetic profiles for 14 compounds of XYP were described by UPLC-MS/MS. A multiple reaction monitoring (MRM) pattern was selected as the quantification model to decrease the matrix effect. The pharmacokinetic study of rats after oral administration of XYP fully verified the effectiveness of the method. It is expected that the present work will provide helpful information on the mechanism (s) of action of XYP and understanding of the pharmacokinetics of multiple active components to further the effective clinical application and safety evaluation of Chinese medicines.

2. Results and Discussion

2.1. Method Validation

2.1.1. Specificity

The representative chromatograms of blank plasma, blank plasma spiked with reference standards and IS, and plasma obtained after the oral administration of XYP are shown in Figure 1. Under the established optimal chromatographic conditions, no significant interfering peaks were observed at the analyte elution times, and no interference occurred between the IS and the 14 analytes.

2.1.2. Linearity and Calibration Curve

The linearity regression equation, correlation coefficients, and linear ranges of the 14 analytes are shown in Table 1. Good linearity was obtained for all analytes. The LLOQ values were appropriate for the quantitative detection of the analytes in pharmacokinetic studies.

2.1.3. Precision and Accuracy

The precision and accuracy of the UPLC-MS/MS method (Table 2) were within acceptable limits. The intra-day accuracy ranged from 90.39–106.52% with a precision of 1.21–11.71%. The inter-day accuracy was 89.93–106.64% with a precision of 2.33–11.55%.

2.1.4. Extraction Recovery and Matrix Effect

Matrix effects and recovery in Table 3 show that no endogenous substance in plasma has a significant effect on the ionization of the analyte. The matrix effects were within an acceptable range 85.17% to 102.69%, and the mean extraction recoveries of the 14 analytes and IS were greater than 82.33%.

2.1.5. Stability

The results of stability experiments (Table 4) showed that no significant degradation occurred. The concentrations of the 14 analytes measured in the stability study were within 7.45% of the initial values. The data indicated that all analytes in rat plasma were stable after storage at −20 °C for 30 days, after three freeze/thaw cycles, and after storage in the auto-sampler (4 °C) for 36 h.

2.1.6. Pharmacokinetic Study

The validated UPLC-MS/MS method was successfully applied to the pharmacokinetic study of 14 compounds in rat plasma after the oral administration of XYP at a dose of 4 g/kg (8-fold the clinical dosage). The mean plasma concentration–time profiles are shown in Figure 2. Pharmacokinetic parameters were calculated and are presented in Table 5.
Paeoniflorin and albiflorin are representative compounds of Radix Paeoniae Alba. The absorption and elimination of paeoniflorin in rat plasma were faster, with more absorption and higher bioavailability than that of albiflorin. Ferulic acid was detected in plasma at 5 min and the plasma concentration reached its peak (Cmax) at 0.2 h (Tmax) for rats (Figure 2), which was very quick and similar to the results found in the literature [28]. The tmax of atractylenolide II and atractylenolide III were similar due to their similar structures. However, atractylenolide II and atractylenolide III showed significant differences in their absorption parameters. The concentrations of the two compounds in XYP extract were similar, but higher levels in the AUC0–t and Cmax of atractylenolide III were observed, compared with atractylenolide II after intragastric administration of XYP extract. The significant differences existed in the absorption of the PK parameters between atractylenolide II and III, which is correspondence with a pharmacokinetic study of atractylenolide II and III in rat plasma after intragastric administration of Baizhufuling extract. This phenomenon may be caused by the higher polarity and slower elimination rate (t1/2) of atractylenolide III [29,30]. Quercetin is a widely used dietary flavonoid, which can be directly absorbed into epithelial cells in the intestine to enter the circulatory system finally. Therefore, it can be more easily and faster absorbed in plasma with tmax of 0.1 h. The pharmacokinetic profile of liquiritin was similar to a previous report [31,32]. Liquiritin and senkyunolide I were rapidly absorbed and eliminated after oral administration. Compared with previous study [33,34], we found that they showed similar pharmacokinetic profile. The study indicated that ligustilide reached a maximum concentration within approximately 0.3 h and the pharmacokinetic profile of ligustilide was accordance with a previous research [35].
As flavonoid aglycones, liquiritigenin, isoliquiritigenin and quercetin reached the second maximal plasma concentrations at about 4 h, 4 h and 8 h. Saikosaponin a, saikosaponin c and glycyrrhizic acid are triterpene glycosides with double peaks in the plasma concentration-time curves as well. This might be attributed to some factors that include enterohepatic recirculation, variable gastric emptying, multiple sites absorption, formulation, etc. The first pass effect of liver and intestine probably contributed to the high concentration double peaks as well [36,37]. Several reasons could explain the double peak phenomenon of flavonoid aglycones. Firstly, the phenomenon maybe caused with enterohepatic recirculation. The enterohepatic circulation of flavonoids has been reported in serval studies. Secondly, the flavonoid glycosides were probably hydrolyzed to flavonoid aglycones by glucose hydrolase in vivo. Thus, second absorption peaks of flavonoid aglycones existed. The double peak plasma concentration-time curves of liquiritigenin, isoliquiritigenin and liquiritin are identical to the literature ones [33,38].

3. Materials and Methods

3.1. Chemicals and Reagents

Standard compounds of albiflorin, paeoniflorin, ferulic acid, senkyunolide I, quercetin, isoliquiritigenin, atractylenolide III, ligustilide, atractylenolide II, liquiritin, liquiritigenin, saikosaponin c, glycyrrhizic acid, saikosaponin a and naringenin as an internal standard (IS) (≥98% purity) were purchased from Chengdu Herbpurify Co., Ltd. (Chengdu, China). The structures of these compounds are shown in Figure 3.
Acetonitrile (HPLC grade) was purchased from Fisher (Fair Lawn, NJ, USA). Ultrapure water with a sensitivity of 18.2 W was prepared from a Milli-Q water purification system (Millipore, Billerica, MA, USA). All other chemicals were analytical grade.

3.2. Instrumentation and Ultraperformance Liquid Chromatography-Tandem Mass Spectrometry (UPLC-MS/MS) Analytical Conditions

Quantitative analysis was conducted on a Waters® Micromass® Quattro Premier™ XE triple-quadrupole tandem mass spectrometer (Waters Corp., Milford, MA, USA). Chromatographic separation was performed on an ACQUITY UPLCTM BEH C18 column (1.7 µm, 50 × 2.1 mm; Waters Corp.) equipped with an ACQUITY UPLC C18 guard column. The column temperature was maintained at 35 °C. The mobile phase consisted of 0.1% formic acid in water (A) and acetonitrile (B). The total flow rate was 0.3 mL/min. Chromatographic separation was achieved using a 14.5-min gradient elution. The gradient conditions of the mobile phase were as follows: 0–4 min, 4–22% (B); 4–5 min, 22–27% (B); 5–10 min, 27–50% (B); and 10–14.5 min, 50–96% (B). The samples were kept at 4 °C in the auto-sampler, and the injection volume was 5 µL.
The mass spectrometer was operated using an electrospray ionization source in positive and negative ion modes. Quantification was performed in positive or negative MRM mode. The MS parameters for nine compounds (albiflorin, paeoniflorin, ferulic acid, senkyunolide I, quercetin, isoliquiritigenin, atractylenolide III, ligustilide, and atractylenolide II) were optimized in the positive mode, whereas the MS parameters for the other five compounds (liquiritin, liquiritigenin, saikosaponin c, glycyrrhizic acid, and saikosaponin a) were optimized under negative ion mode. The parameters of the mass spectrometer were set as follows: capillary voltage, 3.50 kV (+) and 3.0 kV (−); source temperature, 120 °C; and desolvation temperature 300 °C. Argon was used as the collision gas at a pressure of approximately 2.89 × 10−3 mbar. Nitrogen was used as the desolvation and cone gas with flow rates of 600 and 50 L/h, respectively. In Table 6, the cone voltage and collision energy of each compound were optimized. The dwell time was set as 0.2 s. All data collected were processed using Mass Lynx™ NT 4.1 software with the Quan Lynx™ program (Waters, Milford, MA, USA).

3.3. Preparation of Standards and Quality Control (QC) Samples

Stock solutions (100 µg/mL) of albiflorin, paeoniflorin, ferulic acid, senkyunolide I, quercetin, isoliquiritigenin, atractylenolide III, ligustilide, atractylenolide II, liquiritin, liquiritigenin, saikosaponin c, glycyrrhizic acid, saikosaponin a and naringenin (IS) were separately prepared in methanol. The stock solutions of analytes were further mixed and diluted with methanol to yield standard working solutions. The concentrations of the working solutions ranged from 0.46–480 ng/mL for albiflorin, 1.02–1080 ng/mL for paeoniflorin, 2.08–1100 ng/mL for ferulic acid, 2.70–1150 ng/mL for senkyunolide I, 2.15–1085 ng/mL for quercetin, 0.54–485 ng/mL for isoliquiritigenin, 1.05–1105 ng/mL for atractylenolide III, 1.44–1050 ng/mL for ligustilide, 0.51–480 ng/mL for atractylenolide II, 0.53–495 ng/mL for liquiritin, 1.13–1050 ng/mL for liquiritigenin, 0.48–510 ng/mL for saikosaponin c, 0.96–1120 ng/mL for glycyrrhizic acid, and 0.49–510 ng/mL for saikosaponin a. The working solution of IS was prepared by diluting the stock solution to a final concentration of 10 μg/mL using methanol. Three levels of QC samples (low, intermediate and high) were prepared in the same manner. All samples were stored at −20 °C.

3.4. Preparation and Quality Assessment of XYP Extract

The crude plant materials purchased at Tongren Tang. Professor Zhang Ning of Heilongjiang University of Traditional Chinese Medicine identify correct plants by the Chinese Pharmacopoeia 2015. The Identification results were preserved in Heilongjiang University of Traditional Chinese Medicine. The crude powders of Radix Bupleuri (100 g), Radix Angelicae Sinensis (100 g), Radix Paeoniae Alba (100 g), Rhizoma Atractylodis Macrocephalae (100 g), Poria (100 g), Radix Glycyrrhizae (80 g), Herba Menthae (20 g), and Rhizoma Zingiberis Recens (20 g) were mixed together and refluxed with 60% ethanol for 2 h three times. The filtered decoctions are merged, evaporated under reduce pressure at 50 °C and lyophilized with a yield of 26.1%. To calculate the administration dose, the 14 target compounds in XYP extract were determined by UPLC-MS/MS.
The dried extract was dissolved in 70% methanol with a solid–liquid ratio of 30:1 mg/mL (w/v), after centrifugation, supernatant was injected into the detector. Then, the content is calculated using the obtained standard curve. The quantitative result showed the contents of albiflorin, paeoniflorin, ferulic acid, senkyunolide I, quercetin, isoliquiritigenin, atractylenolide III, ligustilide, atractylenolide II, liquiritin, liquiritigenin, saikosaponin c, glycyrrhizic acid, saikosaponin a in XYP extract to be 0.62, 1.7, 5.46, 1.45, 0.70, 0.13, 1.42, 0.14, 1.01, 1.42, 1.23, 0.10, 1.12, 0.09 mg/g, respectively.

3.5. Plasma Sample Preparation

Methanol (1.5 mL), 10 μg/mL IS and 300 μL plasma was mixed, then vortexed for 1 min. After centrifuging at 14,000× g for 10 min, the supernatant was put into another centrifuge tube. The residue was vortexed with another 1.5 mL methanol and centrifuged again, and the two supernatants were combined and evaporated to dryness under a flow of nitrogen gas at 40 °C. The residue was then dissolved in 100 μL methanol and vortexed for 1 min. Finally, the vortexing and centrifugation process was again repeated, and 5 μL of the supernatant was injected into the UPLC-MS/MS system for analysis.

3.6. Method Validation

3.6.1. Specificity

Specificity was assessed by a comparing the chromatograms of blank plasma, blank plasma with the 14 compounds and IS and a plasma sample obtained after the oral administration of XYP.

3.6.2. Linearity and Lower Limit of Quantitation (LLOQ)

The calibration curves were constructed by the plot of the peak area ratios of the analytes versus the IS against the concentrations of the standards and weight coefficient was 1/x2. The LLOQ was defined as the lowest concentration on the standard curve that could be quantitated with an accuracy of 80–120% and a precision (relative standard deviation, RSD, %) value not exceeding 20%.

3.6.3. Precision and Accuracy

Intra- and inter-day accuracy and precision were evaluated from replicate analyses (n = 6) of XYP samples containing different concentrations of analytes (low, intermediate, and high). The results confirm that the proposed method provides satisfactory accuracy and precision for the quantitation of all 14 analytes.

3.6.4. Extraction Recovery and Matrix Effect

The extraction recoveries of the analytes at the three QC levels were determined by comparing the peak areas obtained from 14 extracted QC samples with those obtained from pure reference standards spiked into post-extracted blank rat plasma at the same concentrations. The matrix effects were evaluated by comparing the peak areas obtained from samples for which the extracted matrix was spiked with a standard solution to those obtained from a pure reference standard solution at the same concentrations.

3.5.5. Stability

The stabilities (expressed as relative error, RE, %) of the 14 analytes in plasma were assessed by analyzing the samples with three concentrations (high, intermediate, and low) during the sample storage and processing procedures. The short-term stability was assessed by analyzing the QC samples kept in the auto-sampler (4 °C) for 36 h. To assess long-term stability, the QC samples were stored at −20 °C for 30 days. Freeze-thaw stability was evaluated by subjecting the QC plasma samples to three complete freeze/thaw cycles from −20 °C to room temperature.

3.7. Pharmacokinetic Study

Twenty healthy female Sprague–Dawley rats (300 ± 20 g) were purchased from the Animal Safety Evaluation Center of Heilongjiang University of Chinese Medicine (Heilongjiang, China). All protocols for animal experiments were approved in accordance with the Regulations of Experimental Animal Administration issued by the State Commission of Science and Technology of the People’s Republic of China. The rats were kept under a light/dark cycle of 12 h, within the temperature range of 24 °C ± 2 °C to room temperature, and at a humidity of 50 ± 10%. All rats were fasted for 12 h before experimentation but were allowed to drink freely. XYP extract was dissolved in water, and the rats were administrated orally with 4 g/kg XYP. Blood samples were collected from the suborbital venous lexus at 0, 0.083, 0.17, 0.33, 0.50, 0.75, 1.0, 2.0, 4.0, 8.0, 12 and 24 h into 1.5-mL heparinized polythene tubes before dosing. The blood samples were immediately centrifuged at 4500 rpm for 10 min, and the plasma was stored at −80 °C until analysis. The pharmacokinetic parameters (Cmax, tmax, t1/2, Kel, AUC0–t, AUC0–∞, MRT0–t, MRT0–∞) were calculated with the software Winnolin 3.2.

4. Conclusions

A sensitive and selective UPLC-MS/MS method was developed and applied in a multi-component pharmacokinetic study of XYP administered orally to rats at a dose of 4 g/kg. The developed method can be used to monitor albiflorin, paeoniflorin, ferulic acid, senkyunolide I, quercetin, isoliquiritigenin, atractylenolide III, ligustilide, atractylenolide II, liquiritin, liquiritigenin, saikosaponin c, glycyrrhizic acid and saikosaponin a in rat plasma. The compounds of different structural types (saponins, flavonoids, terpenoids, phenylpropionic acids, coumarin and phthalide) exhibited characteristic pharmacokinetic behaviors. The UPLC-MS/MS method represents the first documented assay for the pharmacokinetic determination of the multiple active ingredients in XYP. This method is expected to be valuable to identify bioactive constituents, elucidate mechanisms of action, and guide the rational clinical use of herbal medicines.

Author Contributions

Conceptualization, F.G. and N.Z.; Data curation, M.X. and Q.X.; Formal analysis, Q.X.; Funding acquisition, F.G.; Investigation, M.X. and H.Z.; Methodology, M.X. and Z.X.; Project administration, M.X. and F.G.; Resources, Q.X., F.G. and N.Z.; Software, H.Z. and M.L.; Supervision, N.Z.; Validation, Z.X. and M.L.; Visualization, H.Z.; Writing—riginal draft, M.X.; Writing—review & editing, F.G.

Funding

This research was funded by the Natural Science Foundation of China (General Program, No. 81673621 and 81673581), Scientific Research Project of Harbin Normal University (No. 14XYG-03), Provincial Universities technological achievements Research and Cultivation fund (TSTAU-C2018020), Heilongjiang applied technology research and development fund (GA17C009).

Acknowledgments

The study was financially supported by the Natural Science Foundation of China (General Program, No. 81673621 and 81673581), Scientific Research Project of Harbin Normal University (No. 14XYG-03), Provincial Universities technological achievements Research and Cultivation fund (TSTAU-C2018020), Heilongjiang applied technology research and development fund (GA17C009).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Geng, F.; Zhang, N.; Fang, H.; Li, J.M.; Zhao, X.; Liu, H.Y. Metabonomic study on protective effect of xiaoyao powder for acute hepatic injury in rats. Zhong Yao Cai 2014, 37, 275–279. [Google Scholar] [PubMed]
  2. Li, X.J.; Ma, Q.Y.; Jiang, Y.M.; Bai, X.H.; Yan, Z.Y.; Liu, Q.; Pan, Q.X.; Liu, Y.Y.; Chen, J.X. Xiaoyaosan exerts anxiolytic-like effects by down-regulating the TNF-α/JAK2-STAT3 pathway in the rat hippocampus. Sci. Rep. 2017, 7, 353. [Google Scholar] [CrossRef] [PubMed]
  3. Man, C.F.; Li, C.; Gong, D.D.; Xu, J.; Fan, Y. Meta-analysis of chinese herbal xiaoyao formula as an adjuvant treatment in relieving depression in chinese patients. Complement Ther. Med. 2014, 22, 362–370. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, J.J.; Li, X.F.; He, S.G.; Hu, L.J.; Guo, J.W.; Huang, X.N.; Hu, J.Q.; Qi, Y.Q.; Chen, B.; Shang, D.W.; et al. Regulation of the kynurenine metabolism pathway by xiaoyao san and the underlying effect in the hippocampus of the depressed rat. J. Ethnopharmacol. 2018, 214, 13–21. [Google Scholar] [CrossRef] [PubMed]
  5. Chinese Pharmacopoeia Commission. Pharmacopoeia of the People’s Republic of China, 2015 ed.; Chinese Medical Science and Technology Press: Beijing, China, 2015; Volume 1, p. 11354. [Google Scholar]
  6. Geng, F.; Zou, T.B.; Wang, Y.Q.; Li, J.M.; Zhang, N.; Ren, Y.D.; Liu, H.Y. UPLC-MS fingerprint of Xiaoyao San. Chin. J. Pharm. Anal. 2013, 33, 1250–1253. [Google Scholar]
  7. Liu, H.; Wang, Y.Y.; Yu, Q.; Li, J.M.; Zhang, N.; Geng, F. GC-MS characteristic chromatograms of xiaoyao powder. Chin. J. Pharm. Anal. 2013, 33, 1507–1517. [Google Scholar]
  8. Zhang, N.; Fang, H.; Wang, X.; Zhang, C.S.; Liu, B.; Zhao, X.; Geng, F. Metabonomic study of intervene effect of xiaoyao powder on rats with hepatic fibrosis. Chin. J. Pharm. Anal. 2014, 34, 588–594. [Google Scholar]
  9. Wang, X.Y.; Ma, X.H.; Li, W.; Chu, Y.; Guo, J.H.; Li, S.M.; Wang, J.M.; Zhang, H.C.; Zhou, S.P.; Zhu, Y.H. Simultaneous determination of five phenolic components and paeoniflorin in rat plasma by liquid chromatography-tandem mass spectrometry and pharmacokinetic study after oral administration of cerebralcare granule (®). J. Pharm. Biomed. Anal. 2013, 86, 82–91. [Google Scholar] [CrossRef] [PubMed]
  10. Wang, R.; Zhang, H.; Wang, Y.Y.; Song, F.X.; Yuan, Y.F. Inhibitory effects of quercetin on the progression of liver fibrosis through the regulation of NF-кB/IкBα, p38 MAPK, and Bcl-2/Bax signaling. Int. Immunopharmacol. 2017, 47, 126–133. [Google Scholar] [CrossRef] [PubMed]
  11. Du, Z.A.; Sun, M.N.; Hu, Z.S. Saikosaponin a ameliorates LPS-Induced acute lung injury in mice. Inflammation 2018, 41, 193–198. [Google Scholar] [CrossRef] [PubMed]
  12. Lee, T.H.; Park, S.; You, M.H.; Lim, J.H.; Min, S.H.; Kim, B.M. A potential therapeutic effect of saikosaponin c as a novel dual-target anti-Alzheimer agent. J. Neurochem. 2016, 136, 1232–1245. [Google Scholar] [CrossRef] [PubMed]
  13. Kuang, X.; Zhou, H.J.; Thorne, A.H.; Chen, X.N.; Li, L.J.; Du, J.R. Neuroprotective effect of ligustilide through induction of α-Secretase processing of both APP and klotho in a mouse model of alzheimer’s disease. Front. Aging Neurosci. 2017, 9, 353. [Google Scholar] [CrossRef] [PubMed]
  14. Luo, N.; Li, Z.; Qian, D.; Qian, Y.; Guo, J.; Duan, J.; Zhu, M. Simultaneous determination of bioactive components of Radix Angelicae Sinensis—Radix Paeoniae Alba herb couple in rat plasma and tissues by UPLC-MS/MS and its application to pharmacokinetics and tissue distribution. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2014, 963, 29–39. [Google Scholar] [CrossRef] [PubMed]
  15. Kim, H.Y.; Park, J.Y.; Lee, K.H.; Lee, D.U.; Kwak, J.H.; Kim, Y.S.; Lee, S.M. Ferulic acid protects against carbon tetrachloride-induced liver injury in mice. Toxicology 2011, 282, 104–111. [Google Scholar] [CrossRef] [PubMed]
  16. Kim, Y.W.; Ki, S.H.; Lee, J.R.; Lee, S.J.; Kim, C.W.; Kim, S.C.; Kim, S.G. Liquiritigenin, an aglycone of liquiritin in Glycyrrhizae radix, prevents acute liver injuries in rats induced by acetaminophen with or without buthionine sulfoximine. Chem. Biol. Interact. 2006, 161, 125–138. [Google Scholar] [CrossRef] [PubMed]
  17. Zhao, Z.; Park, S.M.; Guan, L.; Wu, Y.; Lee, J.R.; Kim, S.C.; Kim, Y.W.; Zhao, R. Isoliquiritigenin attenuates oxidative hepatic damage induced by carbon tetrachloride with or without buthionine sulfoximine. Chem. Biol. Interact. 2015, 225, 13–20. [Google Scholar] [CrossRef] [PubMed]
  18. Zhao, Z.Y.; Wang, W.X.; Guo, H.Z.; Zhou, D.F. Antidepressant-like effect of liquiritin from glycyrrhiza uralensis in chronic variable stress induced depression model rats. Behav. Brain. Res. 2008, 194, 108–113. [Google Scholar] [CrossRef]
  19. Sun, X.; Duan, X.P.; Wang, C.Y.; Liu, Z.H.; Sun, P.Y.; Huo, X.K.; Ma, X.D.; Sun, H.J.; Liu, K.X.; Meng, Q. Protective effects of glycyrrhizic acid against non-alcoholic fatty liver disease in mice. Eur. J. Pharmacol. 2017, 806, 75–82. [Google Scholar] [CrossRef] [PubMed]
  20. Wang, B.; Lian, Y.J.; Dong, X.; Peng, W.; Liu, L.L.; Su, W.J.; Gong, H.; Zhang, T.; Jiang, C.L.; Li, J.S.; et al. Glycyrrhizic acid ameliorates the kynurenine pathway in association with its antidepressant effect. Behav. Brain. Res. 2018. [Google Scholar] [CrossRef] [PubMed]
  21. Qiu, F.; Zhong, X.; Mao, Q.; Huang, Z. The antidepressant-like effects of paeoniflorin in mouse models. Exp. Ther. Med. 2013, 5, 1113–1116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Qiu, F.M.; Zhong, X.M.; Mao, Q.Q.; Huang, Z. Antidepressant-like effects of paeoniflorin on the behavioural, biochemical, and neurochemical patterns of rats exposed to chronic unpredictable stress. Neurosci. Lett. 2013, 541, 209–213. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, Y.L.; Wang, J.X.; Hu, X.X.; Chen, L.; Qiu, Z.K.; Zhao, N.; Yu, Z.D.; Sun, S.Z.; Xu, Y.Y.; Guo, Y.; et al. Antidepressant-like effects of albiflorin extracted from Radix paeoniae Alba. J. Ethnopharmacol. 2016, 179, 9–15. [Google Scholar] [CrossRef] [PubMed]
  24. Hu, Y.Y.; Duan, M.Y.; Liang, S.; Wang, Y.; Feng, Y. Senkyunolide I protects rat brain against focal cerebral ischemia-reperfusion injury by up-regulating p-Erk1/2, Nrf2/HO-1 and inhibiting caspase 3. Brain. Res. 2015, 1605, 39–48. [Google Scholar] [CrossRef] [PubMed]
  25. Fu, X.Q.; Chou, G.X.; Kwan, H.Y.; Tse, A.K.; Zhao, L.H.; Yuen, T.K.; Cao, H.H.; Yu, H.; Chao, X.J.; Su, T.; et al. Inhibition of STAT3 signalling contributes to the antimelanoma action of atractylenolide II. Exp. Dermatol. 2014, 23, 855–864. [Google Scholar] [CrossRef] [PubMed]
  26. Liu, C.; Zhao, H.; Ji, Z.H.; Yu, X.Y. Neuroprotection of atractylenolide III from Atractylodis macrocephalae against glutamate-induced neuronal apoptosis via inhibiting caspase signaling pathway. Neurochem. Res. 2014, 39, 1753–1758. [Google Scholar] [CrossRef] [PubMed]
  27. Zhao, L.S.; Li, F.M. UHPLC-MS strategies and applications for bioanalyses related to pharmacokinetics and drug metabolism. Trends Anal. Chem. 2014, 63, 170–179. [Google Scholar] [CrossRef]
  28. Li, W.X.; Guo, J.M.; Tang, Y.P.; Wang, H.; Huang, M.Y.; Qian, D.W.; Duan, J.A. Pharmacokinetic comparison of ferulic acid in normal and blood deficiency rats after oraladministration of Angelica sinensis, Ligusticum chuanxiong and their combination. Int. J. Mol. Sci. 2012, 13, 3583–3597. [Google Scholar] [CrossRef] [PubMed]
  29. Zhu, Z.M.; Li, H.C.; Luo, J.B. Simultaneous determination of atractylenolide I, II, III by HPLC-MS method and its application to pharmacokinetic study. Pharmacol. Clin. Chin. Mater. Med. 2013, 29, 25–29. [Google Scholar]
  30. Yan, H.; Sun, Y.Y.; Zhang, Q.L.; Yang, M.J.; Wang, X.R.; Wang, Y.; Yu, Z.G.; Zhao, Y.L. Simultaneous determination and pharmacokinetic study of Atractylenolide I, II and III in ratplasma after intragastric administration of Baizhufuling extract and Atractylodis extract by UPLC-MS/MS. J. Chromatogr. B 2015, 993–994, 86–92. [Google Scholar] [CrossRef] [PubMed]
  31. Yang, L.L.; Xiao, N.; Li, X.W.; Fan, Y.; Alolga, R.N.; Sun, X.Y.; Wang, S.L.; Li, P.; Qi, L.W. Pharmacokinetic comparison between quercetin and quercetin 3-O-β-glucuronide in rats by UHPLC-MS/MS. Sci. Rep. 2016, 6, 35460. [Google Scholar] [CrossRef] [PubMed]
  32. Day, A.J.; DuPont, M.S.; Ridley, S.; Rhodes, M.; Rhodes, M.J.C.; Morgan, M.R.A.; Williamson, G. Deglycosylation of flavonoid and isoflavonoid glycosides by human small intestine and liver β-glucosidase activity. FEBS Lett. 1998, 436, 71–75. [Google Scholar] [CrossRef] [Green Version]
  33. Qiao, X.; Ye, M.; Xiang, C.; Wang, Q.; Liu, C.F.; Miao, W.J.; Guo, D.A. Analytical strategy to reveal the in vivo process of multi-component herbal medicine: A pharmacokinetic study of licorice using liquid chromatography coupled with triple quadrupole mass spectrometry. J. Chromatogr. A 2012, 1258, 84–93. [Google Scholar] [CrossRef] [PubMed]
  34. He, C.Y.; Wang, S.; Feng, Y.; Liang, S.; Lin, X.; Xu, D.S.; Ruan, K.F. Pharmacokinetics, tissue distribution and metabolism of senkyunolide I, a major bioactivecomponent in Ligusticum chuanxiong Hort. (Umbelliferae). J. Ethnopharmacol. 2012, 142, 706–713. [Google Scholar] [CrossRef] [PubMed]
  35. Shi, Y.F.; He, L.C.; Wang, S. Determination of ligustilide in rat blood and tissues by capillary gas chromatography/mass spectrometry. Biomed. Chromatogr. 2006, 20, 993–998. [Google Scholar] [CrossRef] [PubMed]
  36. Guan, X.; Wang, X.; Yan, K.; Chu, Y.; Li, S.; Li, W.; Yan, X.; Ma, X.; Zhou, S.; Sun, H.; et al. UFLC-MS/MS determination and pharmacokinetic studies of six Saikosaponins in rat plasma after oral administration of Bupleurum Dropping Pills. J. Pharm. Biomed. Anal. 2016, 124, 288–293. [Google Scholar] [CrossRef] [PubMed]
  37. Xu, C.H.; Wang, P.; Wang, Y.; Yang, Y.; Li, D.H.; Li, H.F.; Sun, S.Q.; Wu, X.Z. Pharmacolcinetic comparisons of two different combinations of Shaoyao-Gancao Decoction in rats: Competing mechanisms between paeoniflorin and glycyrrhetinic acid. J. Ethnopharmacol. 2013, 149, 443–452. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, Z.J.; Zheng, S.R.; Li, L.P.; Jiang, H.D. Metabolism of flavonoids in human: A comprehensive review. Curr. Drug Metab. 2014, 15, 48–61. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of the compounds and herbs are available from the authors.
Figure 1. Representative MRM chromatograms of compounds 1–14 and naringenin (IS) in positive ion mode (red) and negative ion mode (blue). (A): blank plasma; (B): blank plasma spiked with the fifteen analytes and IS; (C): plasma sample1h after oral administration of XYP (4 g/kg) (mean ± SD, n = 6). (1) albiflorin, (2) paeoniflorin, (3) quercetin, (4) ferulic acid, (5) senkyunolide I, (6) isoliquiritigenin, (7) atractylenolide III, (8) ligustilide, (9) atractylenolide II, (10) liquiritin, (11) liquiritigenin, (12) saikosaponin c, (13) glycyrrhizic acid, (14) saikosaponin a, (15) naringenin (IS) in positive ion mode and (16) naringenin (IS) in negative ion mode.
Figure 1. Representative MRM chromatograms of compounds 1–14 and naringenin (IS) in positive ion mode (red) and negative ion mode (blue). (A): blank plasma; (B): blank plasma spiked with the fifteen analytes and IS; (C): plasma sample1h after oral administration of XYP (4 g/kg) (mean ± SD, n = 6). (1) albiflorin, (2) paeoniflorin, (3) quercetin, (4) ferulic acid, (5) senkyunolide I, (6) isoliquiritigenin, (7) atractylenolide III, (8) ligustilide, (9) atractylenolide II, (10) liquiritin, (11) liquiritigenin, (12) saikosaponin c, (13) glycyrrhizic acid, (14) saikosaponin a, (15) naringenin (IS) in positive ion mode and (16) naringenin (IS) in negative ion mode.
Molecules 23 02514 g001
Figure 2. Plasma concentration-time profiles of 14 analytes in rats after oral administration of XYP (4 g/kg) (mean ± SD, n = 6).
Figure 2. Plasma concentration-time profiles of 14 analytes in rats after oral administration of XYP (4 g/kg) (mean ± SD, n = 6).
Molecules 23 02514 g002
Figure 3. Chemical structures of the fourteen analytes and the IS (naringenin).
Figure 3. Chemical structures of the fourteen analytes and the IS (naringenin).
Molecules 23 02514 g003
Table 1. Regression equations, linearity and LLOQ.
Table 1. Regression equations, linearity and LLOQ.
AnalytesRegression EquationCorrelation Coefficient (r2)Linear Range (ng/mL)LLOQ (ng/mL)
Albifloriny = 8.9467x + 43.2760.99960.46–4800.46
Paeonifloriny = 2.8358x + 51.2240.99991.02–10801.02
Ferulic acidy = 1.6548x + 0.56080.99892.08–11002.08
Senkyunolide Iy = 3.4788x − 1.53200.99972.70–11502.70
Quercetiny = 1.9983x + 8.41010.99742.15–10852.15
Isoliquiritigeniny = 44.035x + 25.1880.99930.54–4850.54
Atractylenolide IIIy = 30.767x − 9.54280.99991.05–11051.05
Ligustilidey = 2.3836x + 33.1770.99891.44–10501.44
Atractylenolide IIy = 20.035x + 48.4740.99900.51–4800.51
Liquiritiny = 17.470x + 52.5270.99950.53–4950.53
Liquiritigeniny = 0.3824x + 5.45050.99931.13–10501.13
Saikosaponin cy = 38.923x + 39.8430.99880.48–5100.48
Glycyrrhizic acidy = 47.474x + 24.2250.99940.96–11200.96
Saikosaponin ay = 39.303x + 37.5980.99900.49–5100.49
LLOQ: lower limit of quantification.
Table 2. Intra-day and inter-day precision and accuracy (n = 6).
Table 2. Intra-day and inter-day precision and accuracy (n = 6).
AnalytesNormal Concentration (ng/mL)Intra-Day Inter-Day
Concentration Measured (ng/mL)Precision (RSD%)Accuracy (%)Concentration Measured (ng/mL)Precision (RSD%)Accuracy (%)
Albiflorin3.83.86 ± 0.174.38101.673.66 ± 0.226.0696.18
38.036.66 ± 2.777.5596.4839.20 ± 2.235.70103.16
380.0385.32 ± 23.406.07101.40388.22 ± 15.253.93102.16
Paeoniflorin5.14.66 ± 0.398.3391.344.94 ± 0.254.9896.76
510.0495.77 ± 6.011.2197.21491.59 ± 17.603.5896.39
1020.01032.32 ± 36.043.49101.211065.80 ± 70.416.61104.49
Ferulic acid5.15.10 ± 0.152.89100.965.11 ± 0.132.60101.22
505.0502.84 ± 29.695.9099.57486.81 ± 15.093.1096.40
1010.01037.86 ± 84.168.11102.761039.58 ± 102.419.85102.93
Senkyunolide I4.24.13 ± 0.163.9498.214.08 ± 0.194.6597.02
42.039.15 ± 2.506.3993.2142.49 ± 2.415.67101.17
840.0778.87 ± 46.415.9692.72846.56 ± 35.764.22100.78
Quercetin5.04.95 ± 0.152.9898.154.94 ± 0.132.6298.05
504.0474.88 ± 41.798.8094.22494.74 ± 11.512.3398.16
1008.01005.27 ± 43.454.3299.73980.91 ± 26.412.6997.31
Isoliquiritigenin3.83.71 ± 0.184.9898.893.46 ± 0.164.5592.31
37.539.22 ± 1.654.21104.5939.25 ± 2.025.14104.66
375.0370.56 ± 43.3911.7198.82390.86 ± 16.994.35104.23
Atractylenolide III4.54.19 ± 0.255.9393.194.23 ± 0.419.7393.89
90.093.92 ± 9.8010.43104.3595.97 ± 3.443.58106.64
900.0958.69 ± 59.326.19106.52931.56 ± 48.215.18103.51
Ligustilide5.24.94 ± 0.153.0695.034.86 ± 0.5611.5593.40
104.094.37 ± 8.398.8990.7496.37 ± 4.444.6092.66
1040.0940.05 ± 67.127.1490.39973.42 ± 57.665.9293.60
Atractylenolide II2.42.33 ± 0.145.9197.012.24 ± 0.083.4293.19
48.047.12 ± 1.553.2998.1749.78 ± 1.923.87103.70
480.0469.78 ± 33.547.1497.87458.48 ± 24.185.2795.52
Liquiritin2.11.94 ± 0.094.4894.722.01 ± 0.115.3998.21
41.039.30 ± 1.563.9695.8540.67 ± 3.167.7699.19
410.0383.02 ± 28.287.3893.42391.23 ± 20.065.1395.42
Liquiritigenin4.54.20 ± 0.286.7993.264.05 ± 0.286.9689.93
90.093.48 ± 5.165.52103.8793.71 ± 2.873.06104.12
900.0952.35 ± 52.755.54105.82959.66 ± 71.447.44106.63
Saikosaponin c1.21.16 ± 0.065.5896.251.21 ± 0.065.02101.11
24.022.33 ± 2.099.3793.0223.34 ± 2.088.8997.26
480.0504.04 ± 11.292.24105.01493.36 ± 13.732.78102.78
Glycyrrhizic acid4.64.24 ± 0.348.0392.254.32 ± 0.306.9093.91
460.0430.82 ± 24.475.6893.66434.50 ± 27.536.3494.46
920.0910.12 ± 33.663.7098.93935.77 ± 22.662.42101.71
Saikosaponin a1.31.20 ± 0.065.2094.231.17 ± 0.075.9691.99
25.526.78 ± 1.184.42105.0123.51 ± 2.008.5192.18
510.0491.71 ± 14.572.9696.41496.81 ± 15.443.1197.41
RSD%: expressed as relative standard deviation.
Table 3. Extraction recovery and matrix effect at low, intermediate and high concentration levels (mean ± SD, n = 6).
Table 3. Extraction recovery and matrix effect at low, intermediate and high concentration levels (mean ± SD, n = 6).
AnalyteNormal Concentration (ng/mL)Extraction Recovery (%)Matrix Effect (%)
Albiflorin3.8101.11 ± 2.9095.28 ± 4.49
38101.49 ± 4.9993.33 ± 2.71
38095.48 ± 6.1493.81 ± 4.46
Paeoniflorin5.194.74 ± 4.6392.58 ± 3.26
510101.03 ± 5.8695.36 ± 4.47
102094.73 ± 5.9894.54 ± 3.24
Ferulic acid5.194.23 ± 5.3391.89 ± 4.10
50596.97 ± 3.0086.75 ± 2.97
101099.97 ± 5.0698.31 ± 5.77
Senkyunolide I4.2105.77 ± 6.4297.43 ± 4.98
42103.25 ± 3.0493.14 ± 3.80
840109.75 ± 7.5597.42 ± 5.82
Quercetin588.84 ± 2.4088.56 ± 2.23
50488.58 ± 3.6195.25 ± 6.04
100888.58 ± 3.6186.95 ± 3.81
Isoliquiritigenin3.895.24 ± 6.5794.24 ± 6.16
37.5103.67 ± 5.2097.77 ± 2.90
37597.68 ± 7.3394.35 ± 3.53
Atractylenolide III4.594.92 ± 7.8391.59 ± 6.30
9087.21 ± 4.5088.21 ± 3.10
90095.53 ± 7.3895.77 ± 7.23
Ligustilide5.287.68 ± 5.0987.01 ± 3.67
10485.53 ± 3.5885.79 ± 3.37
104085.48 ± 3.9986.81 ± 3.42
Atractylenolide II2.4101.48 ± 7.0698.42 ± 5.00
4899.79 ± 3.8194.96 ± 7.54
480103.41 ± 7.2598.41 ± 3.62
Liquiritin2.1107.64 ± 7.4898.62 ± 4.87
41104.79 ± 2.5794.62 ± 5.85
410111.77 ± 2.80102.69 ± 7.25
Liquiritigenin4.587.65 ± 4.4187.32 ± 4.43
9099.49 ± 7.9799.49 ± 7.97
90099.49 ± 7.9794.66 ± 4.64
Saikosaponin c1.2100.43 ± 6.7792.73 ± 3.79
2499.35 ± 7.4593.85 ± 3.95
48095.81 ± 8.5395.81 ± 8.53
Glycyrrhizic acid4.682.33 ± 2.9785.17 ± 4.42
46085.67 ± 2.6787.47 ± 2.05
92085.18 ± 3.1786.52 ± 4.03
Saikosaponin a1.393.02 ± 6.7992.69 ± 6.93
25.588.10 ± 4.6887.31 ± 4.75
51090.20 ± 6.2891.13 ± 5.58
Table 4. Stability evaluation results (n = 6).
Table 4. Stability evaluation results (n = 6).
AnalytesSpiked (ng/mL)Auto-Sampler
(4 °C, 36 h)
Long-Term
(−20 °C, 30 Days)
Freeze-Thaw
(−20 °C-Room Temperature)
MeasuredRE (%)MeasuredRE (%)MeasuredRE (%)
Albiflorin3.803.91 ± 0.272.813.76 ± 0.50−1.183.65 ± 0.06−4.04
38.0037.83 ± 2.50−0.4538.87 ± 1.832.2937.00 ± 2.32−2.64
380.00389.79 ± 11.122.58386.55 ± 12.401.72385.16 ± 23.241.36
Paeoniflorin5.105.02 ± 0.18−1.634.99 ± 0.07−2.195.01 ± 0.21−1.80
510.00497.41 ± 11.78−2.47501.59 ± 11.11−1.65494.11 ± 8.68−3.12
1020.001056.41 ± 30.283.571032.46 ± 35.151.221030.65 ± 34.591.04
Ferulic acid5.055.10 ± 0.060.925.10 ± 0.110.895.08 ± 0.110.66
505.00481.27 ± 13.64−4.70491.81 ± 7.89−2.61486.01 ± 17.15−3.76
1010.001000.91 ± 21.30−0.901006.25 ± 27.81−0.371016.19 ± 45.280.61
Senkyunolide I4.204.10 ± 0.13−2.464.09 ± 0.13−2.584.09 ± 0.09−2.58
42.0041.50 ± 3.05−1.2041.33 ± 3.09−1.6041.13 ± 3.11−2.07
840.00834.09 ± 23.38−0.70837.06 ± 26.59−0.35824.21 ± 18.77−1.88
Quercetin5.044.95 ± 0.14−1.854.93 ± 0.13−2.284.94 ± 0.14−1.95
504.00494.39 ± 10.55−1.91493.07 ± 13.07−2.17491.55 ± 18.55−2.47
1008.00990.24 ± 31.42−1.76987.58 ± 36.41−2.03986.94 ± 24.22−2.09
Isoliquiritigenin3.753.75 ± 0.16−0.133.71 ± 0.16−1.023.64 ± 0.34−2.89
37.5037.85 ± 1.420.9438.08 ± 1.501.5537.39 ± 1.90−0.30
375.00371.62 ± 13.67−0.90377.52 ± 13.030.67364.56 ± 15.01−2.78
Atractylenolide III4.504.48 ± 0.16−0.374.41 ± 0.14−2.114.44 ± 0.15−1.26
90.0091.66 ± 3.921.8590.31 ± 2.210.3492.25 ± 3.912.50
900.00920.70 ± 44.642.30919.90 ± 34.372.21925.35 ± 39.892.82
Ligustilide5.205.05 ± 0.13−2.825.02 ± 0.32−3.405.09 ± 0.15−2.08
104.00101.10 ± 4.29−2.7999.70 ± 3.76−4.14102.04 ± 4.00−1.89
1040.001023.55 ± 34.04−1.581028.42 ± 35.42−1.111023.38 ± 30.28−1.60
Atractylenolide II2.402.37 ± 0.27−1.252.49 ± 0.313.612.36 ± 0.10−1.60
48.0046.05 ± 4.17−4.0648.45 ± 6.780.9347.29 ± 1.81−1.48
480.00478.79 ± 24.29−0.25465.14 ± 15.69−3.10478.11 ± 25.41−0.39
Liquiritin2.051.96 ± 0.06−4.391.96 ± 0.06−4.231.99 ± 0.07−2.85
41.0040.59 ± 1.99−1.0140.50 ± 2.82−1.2239.80 ± 1.92−2.93
410.00409.01 ± 8.13−0.24407.90 ± 9.65−0.51409.52 ± 11.42−0.12
Liquiritigenin4.504.50 ± 0.12−0.074.35 ± 0.26−3.414.41 ± 0.10−1.93
90.0090.20 ± 3.790.2289.87 ± 3.18−0.1491.81 ± 3.042.01
900.00910.32 ± 16.621.15919.66 ± 9.212.18912.35 ± 24.551.37
Saikosaponin c1.201.15 ± 0.14−4.031.20 ± 0.09−0.281.19 ± 0.18−1.11
24.0023.48 ± 1.37−2.1524.01 ± 2.900.0423.49 ± 2.06−2.12
480.00492.33 ± 8.812.57491.69 ± 4.902.44491.04 ± 9.822.30
Glycyrrhizic acid4.604.62 ± 0.250.434.57 ± 0.19−0.654.54 ± 0.25−1.23
460.00459.19 ± 22.29−0.18462.84 ± 28.060.62447.49 ± 23.22−2.72
920.00920.55 ± 40.710.06930.77 ± 22.181.17910.12 ± 33.66−1.07
Saikosaponin a1.271.32 ± 0.043.541.27 ± 0.09−0.131.26 ± 0.10−1.05
25.5027.40 ± 3.347.4525.17 ± 3.20−1.2926.19 ± 3.612.72
510.00501.35 ± 16.88−1.70508.81 ± 8.83−0.23486.71 ± 15.18−4.57
RE%: expressed as relative error.
Table 5. The main pharmacokinetic parameters after oral administration of XYP with 4 g/kg. Data are presented as mean ± SD (n = 6).
Table 5. The main pharmacokinetic parameters after oral administration of XYP with 4 g/kg. Data are presented as mean ± SD (n = 6).
AnalytesParameters
tmax (h)Cmax (ng/mL)Kelt1/2 (h)AUC0–t (ng h/mL)AUC0–∞ (ng h/mL)MRT0–t (h)MRT0–∞ (h)
Albiflorin0.39 ± 0.0958.28 ± 27.500.13 ± 0.067.11 ± 4.57127.67 ± 64.94135.02 ± 65.644.18 ± 1.586.34 ± 4.50
Paeoniflorin0.42 ± 0.09586.92 ± 180.660.16 ± 0.064.82 ± 1.71696.90 ± 210.89717.61 ± 227.274.17 ± 1.384.91 ± 1.68
Ferulic acid0.21 ± 0.10632.47 ± 244.040.15 ± 0.055.30 ± 1.851584.14 ± 850.881611.48 ± 852.753.97 ± 0.804.66 ± 0.52
Senkyunolide I0.36 ± 0.13403.26 ± 201.000.18 ± 0.074.75 ± 2.781582.38 ± 985.861616.46 ± 967.015.31 ± 0.726.37 ± 2.39
Quercetin0.10 ± 0.03491.43 ± 167.800.19 ± 0.074.09 ± 1.24766.28 ± 410.22781.69 ± 415.126.40 ± 0.857.03 ± 1.02
Isoliquiritigenin0.39 ± 0.0933.58 ± 9.370.17 ± 0.125.82 ± 3.25156.61 ± 76.73185.17 ± 89.885.98 ± 1.507.81 ± 3.39
Atractylenolide III0.67 ± 0.26229.66 ± 107.940.11 ± 0.056.95 ± 2.651044.70 ± 496.681125.88 ± 487.046.23 ± 1.558.60 ± 3.13
Ligustilide0.31 ± 0.1364.97 ± 42.210.17 ± 0.105.44 ± 3.09194.11 ± 57.72207.32 ± 58.475.63 ± 2.327.47 ± 4.23
Atractylenolide II0.64 ± 0.2922.48 ± 8.040.21 ± 0.073.54 ± 1.2169.53 ± 25.7270.15 ± 25.634.15 ± 0.824.43 ± 0.98
Liquiritin0.39 ± 0.0957.93 ± 36.640.09 ± 0.049.32 ± 5.49163.02 ± 91.45180.94 ± 90.945.70 ± 1.2510.17 ± 6.79
Liquiritigenin0.19 ± 0.0733.17 ± 11.990.12 ± 0.066.43 ± 2.42286.43 ± 144.06328.43 ± 198.168.85 ± 1.0911.49 ± 3.02
Saikosaponin c0.42 ± 0.090.96 ± 0.330.22 ± 0.114.23 ± 2.835.61 ± 1.925.93 ± 2.196.49 ± 1.177.67 ± 2.70
Glycyrrhizic acid0.33 ± 0.1118.65 ± 6.610.12 ± 0.057.63 ± 5.35138.46 ± 56.98156.17 ± 59.5610.06 ± 0.5813.61 ± 5.72
Saikosaponin a0.42 ± 0.096.05 ± 1.300.12 ± 0.036.46 ± 2.5716.26 ± 5.1518.00 ± 6.886.61 ± 1.128.88 ± 3.49
Cmax: the maximum plasma concentration. tmax: time to reach the maximum concentration. t1/2: half-life. AUC: area under the concentration-time curve. Kel: Elimination rate constant.
Table 6. Precursor/product ion pairs and parameters for MRM of compounds used in this study.
Table 6. Precursor/product ion pairs and parameters for MRM of compounds used in this study.
AnalytesCAS No.Retention Time (min)Precursor Ion SpeciesMRM Transitions Precursor Ion → Product Ion (m/z)Cone Voltage (V)Collision Energy (eV)
Albiflorin39011-90-02.94481.50 [M + H]+481.50 → 104.9010.0020.00
Paeoniflorin23180-57-63.23497.90 [M + NH3]+497.90 → 178.9025.0030.00
Ferulic acid1135-24-63.57195.00 [M + H]+195.00 → 145.0025.0020.00
Senkyunolide I94596-28-84.97225.10 [M + H]+225.10 → 207.0010.0020.00
Quercetin117-39-55.10303.20 [M + H]+303.20 → 153.1030.0035.00
Isoliquiritigenin961-29-57.23257.00 [M + H]+257.00 → 136.8025.0025.00
Atractylenolide III73030-71-49.22249.00 [M + H]+249.00 → 231.0015.0010.00
Ligustilide81944-09-410.22191.20 [M + H]+191.20 → 90.9335.0020.00
Atractylenolide II73069-14-410.82233.30 [M + H]+233.30 → 91.0025.0030.00
Liquiritin551-15-53.76417.00 [M − H]417.00 → 255.0035.0020.00
Liquiritigenin578-86-95.30255.00 [M − H]255.00 → 135.0025.0030.00
Saikosaponin c20736-08-77.55971.60 [M + COOH]971.60 → 925.5035.0025.00
Glycyrrhizic acid1405-86-38.14821.00 [M − H]821.00 → 351.0040.0040.00
Saikosaponin a20736-09-88.76825.40 [M + COOH]825.40 → 779.6035.0045.00
Naringenin480-41-14.43273.10 [M + H]+273.10 → 153.1030.0025.00
Naringenin480-41-14.46271.10 [M − H]271.10 → 151.1030.0025.00
MRM: multiple reaction monitoring.

Share and Cite

MDPI and ACS Style

Xu, M.; Xu, Z.; Xu, Q.; Zhang, H.; Liu, M.; Geng, F.; Zhang, N. UPLC-MS/MS Method for the Determination of 14 Compounds in Rat Plasma and Its Application in a Pharmacokinetic Study of Orally Administered Xiaoyao Powder. Molecules 2018, 23, 2514. https://doi.org/10.3390/molecules23102514

AMA Style

Xu M, Xu Z, Xu Q, Zhang H, Liu M, Geng F, Zhang N. UPLC-MS/MS Method for the Determination of 14 Compounds in Rat Plasma and Its Application in a Pharmacokinetic Study of Orally Administered Xiaoyao Powder. Molecules. 2018; 23(10):2514. https://doi.org/10.3390/molecules23102514

Chicago/Turabian Style

Xu, Mingyue, Zhanling Xu, Qingxuan Xu, Hongyue Zhang, Mingyang Liu, Fang Geng, and Ning Zhang. 2018. "UPLC-MS/MS Method for the Determination of 14 Compounds in Rat Plasma and Its Application in a Pharmacokinetic Study of Orally Administered Xiaoyao Powder" Molecules 23, no. 10: 2514. https://doi.org/10.3390/molecules23102514

Article Metrics

Back to TopTop